101
|
The Progestin Receptor Interactome in the Female Mouse Hypothalamus: Interactions with Synaptic Proteins Are Isoform Specific and Ligand Dependent. eNeuro 2017; 4:eN-NWR-0272-17. [PMID: 28955722 PMCID: PMC5605756 DOI: 10.1523/eneuro.0272-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 01/06/2023] Open
Abstract
Progestins bind to the progestin receptor (PR) isoforms, PR-A and PR-B, in brain to influence development, female reproduction, anxiety, and stress. Hormone-activated PRs associate with multiple proteins to form functional complexes. In the present study, proteins from female mouse hypothalamus that associate with PR were isolated using affinity pull-down assays with glutathione S-transferase–tagged mouse PR-A and PR-B. Using complementary proteomics approaches, reverse phase protein array (RPPA) and mass spectrometry, we identified hypothalamic proteins that interact with PR in a ligand-dependent and isoform-specific manner and were confirmed by Western blot. Synaptic proteins, including synapsin-I and synapsin-II, interacted with agonist-bound PR isoforms, suggesting that both isoforms function in synaptic plasticity. In further support, synaptogyrin-III and synapsin-III associated with PR-A and PR-B, respectively. PR also interacted with kinases, including c-Src, mTOR, and MAPK1, confirming phosphorylation as an integral process in rapid effects of PR in the brain. Consistent with a role in transcriptional regulation, PR associated with transcription factors and coactivators in a ligand-specific and isoform-dependent manner. Interestingly, both PR isoforms associated with a key regulator of energy homeostasis, FoxO1, suggesting a novel role for PR in energy metabolism. Because many identified proteins in this PR interactome are synaptic proteins, we tested the hypothesis that progestins function in synaptic plasticity. Indeed, progesterone enhanced synaptic density, by increasing synapsin-I–positive synapses, in rat primary cortical neuronal cultures. This novel combination of RPPA and mass spectrometry allowed identification of PR action in synaptic remodeling and energy homeostasis and reveals unique roles for progestins in brain function and disease.
Collapse
|
102
|
O'Leary KA, Shea MP, Salituro S, Blohm CE, Schuler LA. Prolactin Alters the Mammary Epithelial Hierarchy, Increasing Progenitors and Facilitating Ovarian Steroid Action. Stem Cell Reports 2017; 9:1167-1179. [PMID: 28919264 PMCID: PMC5639259 DOI: 10.1016/j.stemcr.2017.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 01/01/2023] Open
Abstract
Hormones drive mammary development and function and play critical roles in breast cancer. Epidemiologic studies link prolactin (PRL) to increased risk for aggressive cancers that express estrogen receptor α (ERα). However, in contrast to ovarian steroids, PRL actions on the mammary gland outside of pregnancy are poorly understood. We employed the transgenic NRL-PRL model to examine the effects of PRL alone and with defined estrogen/progesterone exposure on stem/progenitor activity and regulatory networks that drive epithelial differentiation. PRL increased progenitors and modulated transcriptional programs, even without ovarian steroids, and with steroids further raised stem cell activity associated with elevated canonical Wnt signaling. However, despite facilitating some steroid actions, PRL opposed steroid-driven luminal maturation and increased CD61+ luminal cells. Our findings demonstrate that PRL can powerfully influence the epithelial hierarchy alone and temper the actions of ovarian steroids, which may underlie its role in the development of breast cancer.
Collapse
Affiliation(s)
- Kathleen A O'Leary
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael P Shea
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Molecular and Environmental Toxicology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Stephanie Salituro
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Courtney E Blohm
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; UW Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI 53792, USA; Molecular and Environmental Toxicology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
103
|
Singh J, Singh R, Gupta P, Rai S, Ganesher A, Badrinarayan P, Sastry GN, Konwar R, Panda G. Targeting progesterone metabolism in breast cancer with l-proline derived new 14-azasteroids. Bioorg Med Chem 2017; 25:4452-4463. [PMID: 28693914 DOI: 10.1016/j.bmc.2017.06.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/05/2017] [Accepted: 06/17/2017] [Indexed: 02/08/2023]
Abstract
Breast cancer cell proliferation is promoted by a variety of mitogenic signals. Classically estrogen is considered as most predominant mitogenic signal in hormone-dependent breast cancer and progesterone is primarily considered to have protective effect. However, it is suggested that some progesterone metabolite may promote breast cancer and progesterone metabolites like 5α-pregnane and 4-pregnene could serve as regulators of estrogen-responsiveness of breast cancer cells. Here, we estimated the potential of alternate targeting of breast cancer via progesterone signalling. l-Proline derived novel 14-azasteroid compounds were screened against MCF-7 and MDA-MB-231 cell lines using MTT assay. In silico studies, cell cycle, Annexin-V-FITC/PI, JC-1 mitochondrial assay, ROS analysis were performed to analyse the impact of hit compound 3b on breast cancer cells. Further, we analysed the impact of hit 3b on the progesterone, its metabolites and enzymes responsible for the conversion of progesterone and its metabolites using ELISA. Data suggests that compound 3b binds and down regulates of 5α-reductase by specifically inhibiting production of progesterone metabolites that are capable of promoting breast cancer proliferation, epithelial mesenchymal transition and migration. This study establishes the proof of concept and generation of new leads for additional targeting of breast cancer.
Collapse
Affiliation(s)
- Jyotsana Singh
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritesh Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Preeti Gupta
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Smita Rai
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Asha Ganesher
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Preethi Badrinarayan
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - G Narahari Sastry
- Centre for Molecular Modelling, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Rituraj Konwar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Chennai 600 113, India.
| | - Gautam Panda
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Chennai 600 113, India.
| |
Collapse
|
104
|
Rižner TL, Thalhammer T, Özvegy-Laczka C. The Importance of Steroid Uptake and Intracrine Action in Endometrial and Ovarian Cancers. Front Pharmacol 2017; 8:346. [PMID: 28674494 PMCID: PMC5474471 DOI: 10.3389/fphar.2017.00346] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/22/2017] [Indexed: 01/06/2023] Open
Abstract
Endometrial and ovarian cancers predominately affect women after menopause, and are more frequently observed in developed countries. These are considered to be hormone-dependent cancers, as steroid hormones, and estrogens in particular, have roles in their onset and progression. After the production of estrogens in the ovary has ceased, estrogen synthesis occurs in peripheral tissues. This depends on the cellular uptake of estrone-sulfate and dehydroepiandrosterone-sulfate, as the most important steroid precursors in the plasma of postmenopausal women. The uptake through transporter proteins, such as those of the organic anion-transporting polypeptide (OATP) and organic anion-transporter (OAT) families, is followed by the synthesis and action of estradiol E2. Here, we provide an overview of the current understanding of this intracrine action of steroid hormones, which depends on the availability of the steroid precursors and transmembrane transporters for precursor uptake, along with the enzymes for the synthesis of E2. The data is also provided relating to the selected transmembrane transporters from the OATP, OAT, SLC51, and ABC-transporter families, and the enzymes involved in the E2-generating pathways in cancers of the endometrium and ovary. Finally, we discuss these transporters and enzymes as potential drug targets.
Collapse
Affiliation(s)
- Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of LjubljanaLjubljana, Slovenia
| | - Theresia Thalhammer
- Department of Pathophysiology and Allergy Research, Centre for Pathophysiology, Infectiology and Immunology, Medical University of ViennaVienna, Austria
| | - Csilla Özvegy-Laczka
- Momentum Membrane Protein Research Group, Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of SciencesBudapest, Hungary
| |
Collapse
|
105
|
Sun L, Guo DH, Liu F, Liu Q, Jiang N, Sun YF, Cai LP, Zheng HX. A MOUSE MODEL OF MAMMARY HYPERPLASIA INDUCED BY ORAL HORMONE ADMINISTRATION. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES 2017. [PMID: 28638887 PMCID: PMC5471472 DOI: 10.21010/ajtcam.v14i4.27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: Mammary hyperplasia is one of the most common benign breast disorders. Although traditional Chinese medicine has a vast experience in the treatment of mammary hyperplasia, it is not accepted widely due to its unclear mechanism. Methods and Materials: To address the mechanism, we developed a mouse model of mammary hyperplasia. We gave mice estradiol valerate tablets and progesterone capsules sequentially for one month by intragastric administration. Results: Mice treated by this method had a series of pathological changes which are similar to those detected in women with mammary hyperplasia, including ectopic level of estradiol and progesterone in serum, hyperplasia of mammary glands and increased expression of ERα and PR. Conclusion: This model will facilitate the mechanical study of traditional medicine on mammary hyperplasia.
Collapse
Affiliation(s)
- Li Sun
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| | - Dong-Hui Guo
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| | - Fei Liu
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| | - Qian Liu
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| | - Ning Jiang
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| | - Yun-Feng Sun
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| | - Li-Ping Cai
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| | - Hong-Xin Zheng
- Molecular Laboratory of TCM, Department of Basic Medicine, Liaoning University of Traditional Chinese Medicine, China
| |
Collapse
|
106
|
Rojas PA, May M, Sequeira GR, Elia A, Alvarez M, Martínez P, Gonzalez P, Hewitt S, He X, Perou CM, Molinolo A, Gibbons L, Abba MC, Gass H, Lanari C. Progesterone Receptor Isoform Ratio: A Breast Cancer Prognostic and Predictive Factor for Antiprogestin Responsiveness. J Natl Cancer Inst 2017; 109:3064537. [PMID: 28376177 DOI: 10.1093/jnci/djw317] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/01/2016] [Indexed: 12/12/2022] Open
Abstract
Background Compelling evidence shows that progestins regulate breast cancer growth. Using preclinical models, we demonstrated that antiprogestins are inhibitory when the level of progesterone receptor isoform A (PR-A) is higher than that of isoform B (PR-B) and that they might stimulate growth when PR-B is predominant. The aims of this study were to investigate ex vivo responses to mifepristone (MFP) in breast carcinomas with different PR isoform ratios and to examine their clinical and molecular characteristics. Methods We performed human breast cancer tissue culture assays (n = 36) to evaluate the effect of MFP on cell proliferation. PR isoform expression was determined by immunoblotting (n = 282). Tumors were categorized as PRA-H (PR-A/PR-B ≥ 1.2) or PRB-H (PR-A/PR-B ≤ 0.83). RNA was extracted for Ribo-Zero-Seq sequencing to evaluate differentially expressed genes. Subtypes and risk scores were predicted using the PAM50 gene set, the data analyzed using The Cancer Genome Atlas RNA-seq gene analysis and other publicly available gene expression data. Tissue microarrays were performed using paraffin-embedded tissues (PRA-H n = 53, PRB-H n = 24), and protein expression analyzed by immunohistochemistry. All statistical tests were two-sided. Results One hundred sixteen out of 222 (52.3%) PR+ tumors were PRA-H, and 64 (28.8%) PRB-H. Cell proliferation was inhibited by MFP in 19 of 19 tissue cultures from PRA-H tumors. A total of 139 transcripts related to proliferative pathways were differentially expressed in nine PRA-H and seven PRB-H tumors. PRB-H and PRA-H tumors were either luminal B or A phenotypes, respectively ( P = .03). PRB-H cases were associated with shorter relapse-free survival (hazard ratio [HR] = 2.70, 95% confidence interval [CI] = 1.71 to 6.20, P = .02) and distant metastasis-free survival (HR = 4.17, 95% CI = 2.18 to 7.97, P < .001). PRB-H tumors showed increased tumor size ( P < .001), Ki-67 levels ( P < .001), human epidermal growth factor receptor 2 expression ( P = .04), high grades ( P = .03), and decreased total PR ( P = .004) compared with PRA-H tumors. MUC-2 ( P < .001) and KRT6A ( P = .02) were also overexpressed in PRB-H tumors. Conclusion The PRA/PRB ratio is a prognostic and predictive factor for antiprogestin responsiveness in breast cancer.
Collapse
Affiliation(s)
- Paola A Rojas
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María May
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gonzalo R Sequeira
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Andrés Elia
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Michelle Alvarez
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula Martínez
- Hospital de Agudos Magdalena V de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Pedro Gonzalez
- Hospital de Agudos Magdalena V de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Stephen Hewitt
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xiaping He
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Charles M Perou
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Alfredo Molinolo
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Luz Gibbons
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Martin C Abba
- CINIBA-CONICET, Escuela de Ciencias Médicas, UNLP, La Plata, Argentina
| | - Hugo Gass
- Hospital de Agudos Magdalena V de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Claudia Lanari
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
107
|
Lindsten T, Hedbrant A, Ramberg A, Wijkander J, Solterbeck A, Eriksson M, Delbro D, Erlandsson A. Effect of macrophages on breast cancer cell proliferation, and on expression of hormone receptors, uPAR and HER-2. Int J Oncol 2017; 51:104-114. [PMID: 28498427 PMCID: PMC5467790 DOI: 10.3892/ijo.2017.3996] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/27/2017] [Indexed: 12/13/2022] Open
Abstract
Malignant tumors, including breast cancers, are frequently infiltrated with innate immune cells and tumor-associated macrophages (TAMs) represent the major inflammatory component in stroma of many tumors. In this study, we examined the immunoreactivity of the macrophage markers CD68 and CD163 as well as the hormone receptors estrogen receptor α (ERα), progesterone receptor (PR), estrogen receptor β1 (ERβ1), human epidermal growth factor receptor 2 (HER-2), matrix metalloproteinase 9 (MMP‑9), urokinase-type plasminogen activator receptor (uPAR) and the proliferations marker Ki67 in 17 breast cancer biopsies. The quantitative score for CD68+ and CD163+ strongly indicate M2 phenotype dominance in the currently investigated biopsies. We found that an increasing level of macrophages was negatively associated with ERα or PR, whereas a positive association was observed for Ki-67 or uPAR. No significant association could be seen between the level of macrophage and HER-2, ERβ1 or MMP-9 expression. Effect of conditioned media (CM) generated from cultured human M1 and M2 macrophage phenotypes were investigated on the proliferation and expression of selected markers in the T47D breast cancer cell line. We found that in contrast to the in vivo situation, in particularly the CM from M1 macrophages decreased the growth and Ki67 expression in T47D, and significantly increased ERβ1 mRNA levels. Moreover, in accordance to the in vivo situation the CM from the macrophages decreased the expression of ERα protein as well as ERα or PR mRNA. In conclusion our results show that macrophages alone have the capability to decrease the tumor cell expression of ERα and PR in vitro. In the tumor environment in vivo macrophages also contribute to an increase in tumor cell expression of uPAR and Ki67, suggesting that macrophages are involved in impairing the prognosis for breast cancer patients.
Collapse
Affiliation(s)
- Therése Lindsten
- Department of Clinical Pathology and Cytology, Central Hospital Karlstad, SE-651 88 Karlstad, Sweden
| | | | - Anna Ramberg
- Department of Clinical Pathology and Cytology, Central Hospital Karlstad, SE-651 88 Karlstad, Sweden
| | - Jonny Wijkander
- Department of Health Sciences, Karlstad University, SE-651 88 Karlstad, Sweden
| | - Anja Solterbeck
- Department of Clinical Pathology and Cytology, Central Hospital Karlstad, SE-651 88 Karlstad, Sweden
| | - Margareta Eriksson
- Department of Clinical Pathology and Cytology, Central Hospital Karlstad, SE-651 88 Karlstad, Sweden
| | - Dick Delbro
- School of Medical Sciences, Örebro University, SE-702 81 Örebro, Sweden
| | - Ann Erlandsson
- Department of Environmental and Life Sciences/Biology, Karlstad University, SE-651 88 Karlstad, Sweden
| |
Collapse
|
108
|
Scavone C, Bonagura AC, Fiorentino S, Cimmaruta D, Cenami R, Torella M, Fossati T, Rossi F. Efficacy and Safety Profile of Diclofenac/Cyclodextrin and Progesterone/Cyclodextrin Formulations: A Review of the Literature Data. Drugs R D 2017; 16:129-40. [PMID: 26939533 PMCID: PMC4875918 DOI: 10.1007/s40268-016-0123-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background According to health technology assessment, patients deserve the best medicine. The development of drugs associated with solubility enhancers, such as cyclodextrins, represents a measure taken in order to improve the management of patients. Different drugs, such as estradiol, testosterone, dexamethasone, opioids, non-steroidal anti-inflammatories (NSAIDs; i.e. diclofenac), and progesterone are associated with cyclodextrins. Products containing the association of diclofenac/cyclodextrins are available for subcutaneous, intramuscular, and intravenous administration in doses that range from 25 to 75 mg. Medicinal products containing the association of progesterone/cyclodextrins are indicated for intramuscular and subcutaneous injection at a dose equal to 25 mg. Objectives and Methods The effects of cyclodextrins have been discussed in the solubility profile and permeability through biological membranes of drug molecules. A literature search was performed in order to give an overview of the pharmacokinetic characteristics, and efficacy and safety profiles of diclofenac/hydroxypropyl-β-cyclodextrin (HPβCD) and progesterone/HPβCD associations. Results The results of more than 20 clinical studies were reviewed. It was suggested that the new diclofenac/HPβCD formulation gives a rapid and effective response to acute pain and, furthermore, has pharmacokinetic and efficacy/safety profiles comparable to other medicinal products not containing cyclodextrins. One of the principal aspects of these new diclofenac formulations is that in lowering the dose (lower than 50 mg) the drugs could be more tolerable, especially in patients with comorbid conditions. Moreover, results of studies investigating the characteristics of progesterone and cyclodextrins showed that the new formulation (progesterone/HPβCD 25 mg solution) has the same bioavailability as other products containing progesterone. It is more rapidly absorbed and allows the achievement of peak plasma concentrations in a shorter time. Finally, the new formulation of progesterone was shown to be safe and not inferior to other products already on the market, with the exception of progesterone administered vaginally. Conclusions As shown by the results of clinical studies presented in this review, the newly approved medicines containing cyclodextrins have been found to be as effective and as well-tolerated as other medicinal products that do not contain cyclodextrins. Moreover, the newly approved lower dose of diclofenac associated with cyclodextrins is consistent with the European Medicines Agency recommendations reported in the revision of the Assessment Report for Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) and Cardiovascular Risk. Finally, the use of cyclodextrins led to significant increases in solubility and bioavailability of drugs, such as diclofenac and progesterone, and improvement in the efficacy and safety of these drugs.
Collapse
Affiliation(s)
- Cristina Scavone
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine, Second University of Naples, Via De Crecchio, 7, Naples, 80138, Italy.
| | - Angela Colomba Bonagura
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine, Second University of Naples, Via De Crecchio, 7, Naples, 80138, Italy
| | - Sonia Fiorentino
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine, Second University of Naples, Via De Crecchio, 7, Naples, 80138, Italy
| | - Daniela Cimmaruta
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine, Second University of Naples, Via De Crecchio, 7, Naples, 80138, Italy
| | - Rosina Cenami
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine, Second University of Naples, Via De Crecchio, 7, Naples, 80138, Italy
| | - Marco Torella
- Department of Women, Child, General and Specialised Surgery, Second University of Naples, Caserta, Italy
| | | | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", School of Medicine, Second University of Naples, Via De Crecchio, 7, Naples, 80138, Italy
| |
Collapse
|
109
|
Wu SP, DeMayo FJ. Progesterone Receptor Signaling in Uterine Myometrial Physiology and Preterm Birth. Curr Top Dev Biol 2017; 125:171-190. [PMID: 28527571 DOI: 10.1016/bs.ctdb.2017.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Myometrium holds the structural integrity for the uterus and generates force for parturition with its primary component, the smooth muscle cells. The progesterone receptor mediates progesterone-dependent signaling and connects to a network of pathways for regulation of contractility and inflammatory responses in myometrium. Dysfunctional progesterone signaling has been linked to pregnancy complications including preterm birth. In the present review, we summarize recent findings on modifiers and effectors of the progesterone receptor signaling. Discussions include novel conceptual discoveries and new development in legacy pathways such as the signal transducers NF-κB, ZEB, microRNA, and the unfolded protein response pathways. We also discuss the impact of progesterone receptor isoform composition and ligand accessibility in modification of the progesterone receptor genomic actions.
Collapse
Affiliation(s)
- San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, United States
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, United States.
| |
Collapse
|
110
|
Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL, Lange CA. Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. J Hematol Oncol 2017; 10:89. [PMID: 28412963 PMCID: PMC5392969 DOI: 10.1186/s13045-017-0462-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/03/2017] [Indexed: 12/18/2022] Open
Abstract
Background Estrogen and progesterone are potent breast mitogens. In addition to steroid hormones, multiple signaling pathways input to estrogen receptor (ER) and progesterone receptor (PR) actions via posttranslational events. Protein kinases commonly activated in breast cancers phosphorylate steroid hormone receptors (SRs) and profoundly impact their activities. Methods To better understand the role of modified PRs in breast cancer, we measured total and phospho-Ser294 PRs in 209 human breast tumors represented on 2754 individual tissue spots within a tissue microarray and assayed the regulation of this site in human tumor explants cultured ex vivo. To complement this analysis, we assayed PR target gene regulation in T47D luminal breast cancer models following treatment with progestin (promegestone; R5020) and antiprogestins (mifepristone, onapristone, or aglepristone) in conditions under which the receptor is regulated by Lys388 SUMOylation (K388 intact) or is SUMO-deficient (via K388R mutation to mimic persistent Ser294 phosphorylation). Selected phospho-PR-driven target genes were validated by qRT-PCR and following RUNX2 shRNA knockdown in breast cancer cell lines. Primary and secondary mammosphere assays were performed to implicate phospho-Ser294 PRs, epidermal growth factor signaling, and RUNX2 in breast cancer stem cell biology. Results Phospho-Ser294 PR species were abundant in a majority (54%) of luminal breast tumors, and PR promoter selectivity was exquisitely sensitive to posttranslational modifications. Phospho-PR expression and target gene programs were significantly associated with invasive lobular carcinoma (ILC). Consistent with our finding that activated phospho-PRs undergo rapid ligand-dependent turnover, unique phospho-PR gene signatures were most prevalent in breast tumors clinically designated as PR-low to PR-null (luminal B) and included gene sets associated with cancer stem cell biology (HER2, PAX2, AHR, AR, RUNX). Validation studies demonstrated a requirement for RUNX2 in the regulation of selected phospho-PR target genes (SLC37A2). In vitro mammosphere formation assays support a role for phospho-Ser294-PRs via growth factor (EGF) signaling as well as RUNX2 as potent drivers of breast cancer stem cell fate. Conclusions We conclude that PR Ser294 phosphorylation is a common event in breast cancer progression that is required to maintain breast cancer stem cell fate, in part via cooperation with growth factor-initiated signaling pathways and key phospho-PR target genes including SLC37A2 and RUNX2. Clinical measurement of phosphorylated PRs should be considered a useful marker of breast tumor stem cell potential. Alternatively, unique phospho-PR target gene sets may provide useful tools with which to identify patients likely to respond to selective PR modulators that block PR Ser294 phosphorylation as part of rational combination (i.e., with antiestrogens) endocrine therapies designed to durably block breast cancer recurrence. Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0462-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Todd P Knutson
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Thu H Truong
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Shihong Ma
- Department of Urology, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, J8.130C, Dallas, TX, 75390-9110, USA
| | - Nicholas J Brady
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Megan E Sullivan
- Department of Pathology, Evanston Hospital, University of Chicago, NorthShore University HealthSystem, Evanston, IL, 60201, USA
| | - Ganesh Raj
- Department of Urology, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, J8.130C, Dallas, TX, 75390-9110, USA
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Carol A Lange
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
111
|
Interactions between dietary acrylamide intake and genes for ovarian cancer risk. Eur J Epidemiol 2017; 32:431-441. [PMID: 28391539 PMCID: PMC5506210 DOI: 10.1007/s10654-017-0244-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/30/2017] [Indexed: 01/03/2023]
Abstract
Some epidemiological studies observed a positive association between dietary acrylamide intake and ovarian cancer risk but the causality needs to be substantiated. By analyzing gene-acrylamide interactions for ovarian cancer risk for the first time, we aimed to contribute to this. The prospective Netherlands Cohort Study on diet and cancer includes 62,573 women, aged 55–69 years. At baseline in 1986, a random subcohort of 2589 women was sampled from the total cohort for a case cohort analysis approach. Dietary acrylamide intake of subcohort members and ovarian cancer cases (n = 252, based on 20.3 years of follow-up) was assessed with a food frequency questionnaire. We selected single nucleotide polymorphisms (SNPs) in genes in acrylamide metabolism and in genes involved in the possible mechanisms of acrylamide-induced carcinogenesis (effects on sex steroid systems, oxidative stress and DNA damage). Genotyping was done on DNA from toenails through Agena’s MassARRAY iPLEX platform. Multiplicative interaction between acrylamide intake and SNPs was assessed with Cox proportional hazards analysis. Among the results for 57 SNPs and 2 gene deletions, there were no statistically significant interactions between acrylamide and gene variants after adjustment for multiple testing. However, there were several nominally statistically significant interactions between acrylamide intake and SNPs in the HSD3B1/B2 gene cluster: (rs4659175 (p interaction = 0.04), rs10923823 (p interaction = 0.06) and its proxy rs7546652 (p interaction = 0.05), rs1047303 (p interaction = 0.005), and rs6428830 (p interaction = 0.05). Although in need of confirmation, results of this study suggest that acrylamide may cause ovarian cancer through effects on sex hormones.
Collapse
|
112
|
Chuffa LGDA, Lupi-Júnior LA, Costa AB, Amorim JPDA, Seiva FRF. The role of sex hormones and steroid receptors on female reproductive cancers. Steroids 2017; 118:93-108. [PMID: 28041951 DOI: 10.1016/j.steroids.2016.12.011] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/10/2016] [Accepted: 12/24/2016] [Indexed: 02/08/2023]
Abstract
Sex steroids have been widely described to be associated with a number of human diseases, including hormone-dependent tumors. Several studies have been concerned about the factors regulating the availability of sex steroids and its importance in the pathophysiological aspects of the reproductive cancers in women. In premenopausal women, large fluctuations in the concentration of circulating estradiol (E2) and progesterone (P4) orchestrate many events across the menstrual cycle. After menopause, the levels of circulating E2 and P4 decline but remain at high concentration in the peripheral tissues. Notably, there is a strong relationship between circulating sex hormones and female reproductive cancers (e.g. ovarian, breast, and endometrial cancers). These hormones activate a number of specific signaling pathways after binding either to estrogen receptors (ERs), especially ERα, ERα36, and ERβ or progesterone receptors (PRs). Importantly, the course of the disease will depend on particular transactivation pathway. Identifying ER- or PR-positive tumors will benefit patients in terms of proper endocrine therapy. Based on hormonal responsiveness, effective prevention methods for ovarian, breast, and endometrial cancers represent a special opportunity for women at risk of malignancies. Hormone replacement therapy (HRT) might significantly increase the risk of these cancer types, and endocrine treatments targeting ER signaling may be helpful against E2-dependent tumors. This review will present the role of sex steroids and their receptors associated with the risk of developing female reproductive cancers, with emphasis on E2 levels in pre and postmenopausal women. In addition, new therapeutic strategies for improving the survival rate outcomes in women will be addressed.
Collapse
Affiliation(s)
| | - Luiz Antonio Lupi-Júnior
- Department of Anatomy, IBB/UNESP, Institute of Biosciences of Botucatu, Univ. Estadual Paulista, SP, Brazil
| | - Aline Balandis Costa
- Department of Nursing, UENP/CLM - Universidade Estadual do Norte do Paraná, PR, Brazil
| | | | | |
Collapse
|
113
|
Hansberg-Pastor V, González-Arenas A, Camacho-Arroyo I. CCAAT/enhancer binding protein β negatively regulates progesterone receptor expression in human glioblastoma cells. Mol Cell Endocrinol 2017; 439:317-327. [PMID: 27663075 DOI: 10.1016/j.mce.2016.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/29/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
Many progesterone (P4) actions are mediated by its intracellular receptor (PR), which has two isoforms (PR-A and PR-B) differentially transcribed from separate promoters of a single gene. In glioblastomas, the most frequent and aggressive brain tumors, PR-B is the predominant isoform. In an in silico analysis we showed putative CCAAT/Enhancer Binding Protein (C/EBP) binding sites at PR-B promoter. We evaluated the role of C/EBPβ in PR-B expression regulation in glioblastoma cell lines, which expressed different ratios of PR and C/EBPβ isoforms (LAP1, LAP2, and LIP). ChIP assays showed a significant basal binding of C/EBPβ, specific protein 1 (Sp1) and estrogen receptor alpha (ERα) to PR-B promoter. C/EBPβ knockdown increased PR-B expression and treatment with estradiol (E2) reduced C/EBPβ binding to the promoter and up-regulated PR-B expression. P4 induced genes were differently regulated when CEBP/β was silenced. These data show that C/EBPβ negatively regulates PR-B expression in glioblastoma cells.
Collapse
Affiliation(s)
- Valeria Hansberg-Pastor
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México (UNAM), Mexico
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, UNAM, Ciudad de México, Mexico.
| |
Collapse
|
114
|
Salaün H, Thariat J, Vignot M, Merrouche Y, Vignot S. Obésité et cancer. Bull Cancer 2017; 104:30-41. [PMID: 28007295 DOI: 10.1016/j.bulcan.2016.11.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 12/15/2022]
|
115
|
Epidemiology of Endometrial Carcinoma: Etiologic Importance of Hormonal and Metabolic Influences. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 943:3-46. [PMID: 27910063 DOI: 10.1007/978-3-319-43139-0_1] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Endometrial carcinoma is the most common gynecologic cancer in developed nations, and the annual incidence is projected to increase, secondary to the high prevalence of obesity, a strong endometrial carcinoma risk factor. Although endometrial carcinomas are etiologically, biologically, and clinically diverse, hormonal and metabolic mechanisms are particularly strongly implicated in the pathogenesis of endometrioid carcinoma, the numerically predominant subtype. The centrality of hormonal and metabolic disturbances in the pathogenesis of endometrial carcinoma, combined with its slow development from well-characterized precursors in most cases, offers a substantial opportunity to reduce endometrial carcinoma mortality through early detection, lifestyle modification, and chemoprevention. In this chapter, we review the epidemiology of endometrial carcinoma, emphasizing theories that link risk factors for these tumors to hormonal and metabolic mechanisms. Future translational research opportunities related to prevention are discussed.
Collapse
|
116
|
Goncharov AI, Maslakova AA, Polikarpova AV, Bulanova EA, Guseva AA, Morozov IA, Rubtsov PM, Smirnova OV, Shchelkunova TA. Progesterone inhibits proliferation and modulates expression of proliferation-Related genes in classical progesterone receptor-negative human BxPC3 pancreatic adenocarcinoma cells. J Steroid Biochem Mol Biol 2017; 165:293-304. [PMID: 27449817 DOI: 10.1016/j.jsbmb.2016.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 03/03/2016] [Accepted: 07/18/2016] [Indexed: 01/07/2023]
Abstract
Recent studies suggest that progesterone may possess anti-tumorigenic properties. However, a growth-modulatory role of progestins in human cancer cells remains obscure. With the discovery of a new class of membrane progesterone receptors (mPRs) belonging to the progestin and adipoQ receptor gene family, it becomes important to study the effect of this hormone on proliferation of tumor cells that do not express classical nuclear progesterone receptors (nPRs). To identify a cell line expressing high levels of mPRs and lacking nPRs, we examined mRNA levels of nPRs and three forms of mPRs in sixteen human tumor cell lines of different origin. High expression of mPR mRNA has been found in pancreatic adenocarcinoma BxPC3 cells, while nPR mRNA has not been detected in these cells. Western blot analysis confirmed these findings at the protein level. We revealed specific binding of labeled progesterone in these cells with affinity constant similar to that of human mPR expressed in yeast cells. Progesterone at high concentration of 20 μM significantly reduced the mRNA levels of proliferation markers Ki67 and PCNA, as well as of cyclin D1, and increased the mRNA levels of cyclin dependent kinase inhibitors p21 and p27. Progesterone (1 μM and 20 μM) significantly inhibited proliferative activity of BxPC3 cells. These results point to anti-proliferative effects of the progesterone high concentrations on BxPC3 cells and suggest that activation of mPRs may mediate this action. Our data are a starting point for further investigations regarding the application of progesterone in pancreatic cancer.
Collapse
Affiliation(s)
- Alexey I Goncharov
- Lomonosov Moscow State University, Biological Faculty, Lenin Hills, 1/12, Moscow 119234, Russia
| | - Aitsana A Maslakova
- Lomonosov Moscow State University, Biological Faculty, Lenin Hills, 1/12, Moscow 119234, Russia
| | - Anna V Polikarpova
- Lomonosov Moscow State University, Biological Faculty, Lenin Hills, 1/12, Moscow 119234, Russia
| | - Elena A Bulanova
- ChemRar High-Tech Center, 2a-1 Rabochaya St., Khimki, Moscow Oblast 141400 Russia
| | - Alexandra A Guseva
- Lomonosov Moscow State University, Biological Faculty, Lenin Hills, 1/12, Moscow 119234, Russia
| | - Ivan A Morozov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov St., Moscow, 119991 Russia
| | - Petr M Rubtsov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov St., Moscow, 119991 Russia
| | - Olga V Smirnova
- Lomonosov Moscow State University, Biological Faculty, Lenin Hills, 1/12, Moscow 119234, Russia
| | - Tatiana A Shchelkunova
- Lomonosov Moscow State University, Biological Faculty, Lenin Hills, 1/12, Moscow 119234, Russia.
| |
Collapse
|
117
|
Sikora MJ. Family Matters: Collaboration and Conflict Among the Steroid Receptors Raises a Need for Group Therapy. Endocrinology 2016; 157:4553-4560. [PMID: 27835038 PMCID: PMC5133350 DOI: 10.1210/en.2016-1778] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antiestrogen therapies targeting the function of estrogen receptor (ER) have been the cornerstone of therapy for ER+ breast cancer for decades. However, as long as these therapies have been in use, it has also been evident that response to antiestrogen therapy is not based solely on ER expression but that other factors modify breast cancer antiestrogen response. Such factors may include ER's relatives in the steroid hormone receptor (HR) family, androgen receptor (AR), progesterone receptor (PR), glucocorticoid receptor (GR), and mineralocorticoid receptor (MR). A series of recent studies has demonstrated that these HRs are not bystanders in ER signaling but rather can alter ER genomic binding and subsequent control of target gene expression. For example, PR and GR may "reprogram" ER binding to DNA toward PR/GR sites; androgen receptor may reverse ER gene regulation functions or regulate ER DNA binding. Accordingly, modulation of HR function concurrently with antiestrogen therapy can either improve antiestrogen response or mediate antiestrogen resistance. This highlights the critical need to better understand how other HRs influence ER function, in particular in the context of antiestrogen therapy. This review discusses recent insights into the mechanisms by which HRs can modify ER function and antiestrogen response, as well as pharmacological implications for antiestrogen therapies and potential combined endocrine therapies.
Collapse
Affiliation(s)
- Matthew J Sikora
- Department of Pathology, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
118
|
Soini T, Hurskainen R, Grénman S, Mäenpää J, Paavonen J, Pukkala E. Impact of levonorgestrel-releasing intrauterine system use on the cancer risk of the ovary and fallopian tube. Acta Oncol 2016; 55:1281-1284. [PMID: 27148621 DOI: 10.1080/0284186x.2016.1175660] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Levonorgestrel-releasing intrauterine system (LNG-IUS) is used for contraception and heavy menstrual bleeding. A long-term hormone therapy can modify the risk of gynecologic cancers. Little is known about the impact of LNG-IUS use on the risk for invasive and borderline ovarian tumor subtypes or for primary fallopian tube carcinoma. We examined the associations of LNG-IUS use with these tumors. MATERIAL AND METHODS We identified from the national Medical Reimbursement Registry of Finland the women aged 30-49 years who had used LNG-IUS for menorrhagia in 1994-2007, and from the Finnish Cancer Registry ovarian cancers and primary fallopian tube carcinomas diagnosed before the age of 55 and by the end of 2013. RESULTS A total of 77 invasive ovarian cancers and seven primary fallopian tube carcinoma cases were diagnosed in a cohort of 93 843 LNG-IUS users during the follow-up of 1 083 126 women-years. The LNG-IUS users had decreased risk for both invasive ovarian cancer [standardized incidence ratio (SIR) 0.59, 95% confidence interval (CI) 0.47-0.73] and for borderline ovarian tumors (SIR 0.76, 95% CI 0.57-0.99) as compared to the background population. The risk of primary fallopian tube carcinoma was not increased (SIR 1.22, 95% CI 0.49-2.50). Decreased risks for mucinous (SIR 0.49, 95% CI 0.24-0.87), endometrioid (SIR 0.55, 95% CI 0.28-0.98), and serous ovarian carcinomas (SIR 0.75, 95% CI 0.55-0.99) were seen in LNG-IUS users. CONCLUSIONS LNG-IUS use associated with decreased risk for both invasive and borderline ovarian tumors. The incidence of primary fallopian tube carcinoma did not significantly differ between LNG-IUS users and the background population.
Collapse
Affiliation(s)
- Tuuli Soini
- Department of Obstetrics and Gynecology, Hyvinkää Hospital, Hyvinkää, Finland
| | - Ritva Hurskainen
- Department of Obstetrics and Gynecology, Hyvinkää Hospital, Hyvinkää, Finland
| | - Seija Grénman
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku, Finland
- University of Turku, Turku, Finland
| | - Johanna Mäenpää
- School of Medicine, University of Tampere, University of Tampere, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland
| | - Jorma Paavonen
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
- University of Helsinki, Helsinki, Finland
| | - Eero Pukkala
- School of Health Sciences, University of Tampere, University of Tampere, Tampere, Finland
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| |
Collapse
|
119
|
Valadez-Cosmes P, Vázquez-Martínez ER, Cerbón M, Camacho-Arroyo I. Membrane progesterone receptors in reproduction and cancer. Mol Cell Endocrinol 2016; 434:166-75. [PMID: 27368976 DOI: 10.1016/j.mce.2016.06.027] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/15/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
Progesterone is a sexual steroid hormone that has a critical role in reproductive processes in males and females of several species, including humans. Furthermore, progesterone has been associated with pathological diseases such as breast, gynecological and brain cancer, regulating cell proliferation, apoptosis, and metastasis. In the past, progesterone actions were thought to be only mediated by its intracellular receptor (PR). However, recent evidence has demonstrated that membrane progesterone receptors (mPRs) mediate most of the non-classical progesterone actions. The role of the different mPRs subtypes in progesterone effects in reproduction and cancer is an emerging and exciting research area. Here we review studies to date regarding mPRs role in reproduction and cancer and discuss their functions and clinical relevance, suggesting mPRs as putative pharmacological targets and disease markers in cancer and diseases associated with reproduction.
Collapse
Affiliation(s)
- Paulina Valadez-Cosmes
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Edgar Ricardo Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
120
|
Mao C, Livezey M, Kim JE, Shapiro DJ. Antiestrogen Resistant Cell Lines Expressing Estrogen Receptor α Mutations Upregulate the Unfolded Protein Response and are Killed by BHPI. Sci Rep 2016; 6:34753. [PMID: 27713477 PMCID: PMC5054422 DOI: 10.1038/srep34753] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 09/15/2016] [Indexed: 12/13/2022] Open
Abstract
Outgrowth of metastases expressing ERα mutations Y537S and D538G is common after endocrine therapy for estrogen receptor α (ERα) positive breast cancer. The effect of replacing wild type ERα in breast cancer cells with these mutations was unclear. We used the CRISPR-Cas9 genome editing system and homology directed repair to isolate and characterize 14 T47D cell lines in which ERαY537S or ERαD538G replace one or both wild-type ERα genes. In 2-dimensional, and in quantitative anchorage-independent 3-dimensional cell culture, ERαY537S and ERαD538G cells exhibited estrogen-independent growth. A progestin further increased their already substantial proliferation in micromolar 4-hydroxytamoxifen and fulvestrant/ICI 182,780 (ICI). Our recently described ERα biomodulator, BHPI, which hyperactivates the unfolded protein response (UPR), completely blocked proliferation. In ERαY537S and ERαD538G cells, estrogen-ERα target genes were constitutively active and partially antiestrogen resistant. The UPR marker sp-XBP1 was constitutively activated in ERαY537S cells and further induced by progesterone in both cell lines. UPR-regulated genes associated with tamoxifen resistance, including the oncogenic chaperone BiP/GRP78, were upregulated. ICI displayed a greater than 2 fold reduction in its ability to induce ERαY537S and ERαD538G degradation. Progestins, UPR activation and perhaps reduced ICI-stimulated ERα degradation likely contribute to antiestrogen resistance seen in ERαY537S and ERαD538G cells.
Collapse
Affiliation(s)
- Chengjian Mao
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Mara Livezey
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ji Eun Kim
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - David J Shapiro
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
121
|
Stires H, Saboya M, Globerman SP, Cohick WS. Peroral Estradiol Is Sufficient to Induce Carcinogen-Induced Mammary Tumorigenesis in Ovariectomized Rats without Progesterone. PLoS One 2016; 11:e0162662. [PMID: 27611094 PMCID: PMC5017759 DOI: 10.1371/journal.pone.0162662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 08/28/2016] [Indexed: 12/02/2022] Open
Abstract
A role for estrogens in breast cancer is widely accepted, however, recent evidence highlights that timing and exposure levels are important in determining whether they elicit harmful versus beneficial effects. The rat chemical carcinogen model has been widely used to study the effects of estrogens but conclusions on the levels that lead to tumor development and an absolute requirement for progesterone (P4) are lacking. A newer method of hormone administration mixes hormones with nut butter for peroral consumption allowing for a less stressful method of long-term administration with lower spikes in serum estradiol (E2) levels. The present study was designed to determine if estrogens alone at a physiological dose can drive carcinogen-induced tumors in ovariectomized (OVX) rats or if P4 is also required using this method of hormone administration. Short-term studies were conducted to determine the dose of estrogen (E) that would lead to increased uterine weight following OVX. Subsequently, rats were OVX on postnatal day (PND) 40 then treated daily with E (600 μg/kg/day), P4 (15 mg/kg/day), or the combination. On PND 50, all rats were injected with nitrosomethylurea to induce mammary tumors. Uterine weights, body weights, and serum E2 levels were measured to demonstrate the efficacy of the method for increasing E2 levels during long-term treatment. After 26 weeks, tumor incidence was similar in Sham, E, and E + P4 animals indicating that E was sufficient to induce tumorigenesis when hormone levels were normalized by this method. This study demonstrates peroral administration can be used in long-term studies to elucidate relationships between different types and levels of steroid hormones.
Collapse
Affiliation(s)
- Hillary Stires
- Department of Animal Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States of America
| | - Mariana Saboya
- Department of Animal Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States of America
| | - Samantha P. Globerman
- Department of Animal Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States of America
| | - Wendie S. Cohick
- Department of Animal Science, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States of America
- * E-mail:
| |
Collapse
|
122
|
Asa C. Weighing the options for limiting surplus animals. Zoo Biol 2016; 35:183-6. [DOI: 10.1002/zoo.21293] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 04/25/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Cheryl Asa
- AZA Reproductive Management Center; Saint Louis Zoo; St. Louis Missouri
| |
Collapse
|
123
|
Yang X, Zhang W, Chen Y, Li Y, Sun L, Liu Y, Liu M, Yu M, Li X, Han J, Duan Y. Activation of Peroxisome Proliferator-activated Receptor γ (PPARγ) and CD36 Protein Expression: THE DUAL PATHOPHYSIOLOGICAL ROLES OF PROGESTERONE. J Biol Chem 2016; 291:15108-18. [PMID: 27226602 DOI: 10.1074/jbc.m116.726737] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Indexed: 12/27/2022] Open
Abstract
Progesterone or its analog, one of components of hormone replacement therapy, may attenuate the cardioprotective effects of estrogen. However, the underlying mechanisms have not been fully elucidated. Expression of CD36, a receptor for oxidized LDL (oxLDL) that enhances macrophage/foam cell formation, is activated by the transcription factor peroxisome proliferator-activated receptor γ (PPARγ). CD36 also functions as a fatty acid transporter to influence fatty acid metabolism and the pathophysiological status of several diseases. In this study, we determined that progesterone induced macrophage CD36 expression, which is related to progesterone receptor (PR) activity. Progesterone enhanced cellular oxLDL uptake in a CD36-dependent manner. Mechanistically, progesterone increased PPARγ expression and PPARγ promoter activity in a PR-dependent manner and the binding of PR with the progesterone response element in the PPARγ promoter. Specific deletion of macrophage PPARγ (MφPPARγ KO) expression in mice abolished progesterone-induced macrophage CD36 expression and cellular oxLDL accumulation. We also determined that, associated with gestation and increased serum progesterone levels, CD36 and PPARγ expression in mouse adipose tissue, skeletal muscle, and peritoneal macrophages were substantially activated. Taken together, our study demonstrates that progesterone can play dual pathophysiological roles by activating PPARγ expression, in which progesterone increases macrophage CD36 expression and oxLDL accumulation, a negative effect on atherosclerosis, and enhances the PPARγ-CD36 pathway in adipose tissue and skeletal muscle, a protective effect on pregnancy.
Collapse
Affiliation(s)
| | | | - Yuanli Chen
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China School of Medicine, and
| | - Yan Li
- From the College of Life Sciences
| | - Lei Sun
- From the College of Life Sciences
| | - Ying Liu
- From the College of Life Sciences
| | | | - Miao Yu
- From the College of Life Sciences
| | | | - Jihong Han
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071, China and
| | - Yajun Duan
- the College of Biomedical Engineering, Hefei University of Technology, Hefei 230000, China College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Biotherapy, Nankai University, Tianjin 300071, China and
| |
Collapse
|
124
|
Rui W, Zou Y, Lee J, Nambiar SM, Lin J, Zhang L, Yang Y, Dai G. Nuclear Factor Erythroid 2-Related Factor 2 Deficiency Results in Amplification of the Liver Fat-Lowering Effect of Estrogen. J Pharmacol Exp Ther 2016; 358:14-21. [PMID: 27189962 DOI: 10.1124/jpet.115.231316] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/05/2016] [Indexed: 12/16/2022] Open
Abstract
Transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates multiple biologic processes, including hepatic lipid metabolism. Estrogen exerts actions affecting energy homeostasis, including a liver fat-lowering effect. Increasing evidence indicates the crosstalk between these two molecules. The aim of this study was to evaluate whether Nrf2 modulates estrogen signaling in hepatic lipid metabolism. Nonalcoholic fatty liver disease (NAFLD) was induced in wild-type and Nrf2-null mice fed a high-fat diet and the liver fat-lowering effect of exogenous estrogen was subsequently assessed. We found that exogenous estrogen eliminated 49% and 90% of hepatic triglycerides in wild-type and Nrf2-null mice with NAFLD, respectively. This observation demonstrates that Nrf2 signaling is antagonistic to estrogen signaling in hepatic fat metabolism; thus, Nrf2 absence results in striking amplification of the liver fat-lowering effect of estrogen. In addition, we found the association of trefoil factor 3 and fatty acid binding protein 5 with the liver fat-lowering effect of estrogen. In summary, we identified Nrf2 as a novel and potent inhibitor of estrogen signaling in hepatic lipid metabolism. Our finding may provide a potential strategy to treat NAFLD by dually targeting Nrf2 and estrogen signaling.
Collapse
Affiliation(s)
- Wenjuan Rui
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| | - Yuhong Zou
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| | - Joonyong Lee
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| | - Shashank Manohar Nambiar
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| | - Jingmei Lin
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| | - Linjie Zhang
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| | - Yan Yang
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| | - Guoli Dai
- Department of Pharmacology and Immunology, Anhui Medical University, Hefei, China (W.R., L.Z., Y.Y.); Department of Biology, School of Science, Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana (W.R., Y.Z., S.M.N., G.D.); and Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana (J.L.)
| |
Collapse
|
125
|
Leehy KA, Regan Anderson TM, Daniel AR, Lange CA, Ostrander JH. Modifications to glucocorticoid and progesterone receptors alter cell fate in breast cancer. J Mol Endocrinol 2016; 56:R99-R114. [PMID: 26831511 PMCID: PMC7256961 DOI: 10.1530/jme-15-0322] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/16/2015] [Indexed: 12/21/2022]
Abstract
Steroid hormone receptors (SRs) are heavily posttranslationally modified by the reversible addition of a variety of molecular moieties, including phosphorylation, acetylation, methylation, SUMOylation, and ubiquitination. These rapid and dynamic modifications may be combinatorial and interact (i.e. may be sequential, complement, or oppose each other), creating a vast array of uniquely modified receptor subspecies that allow for diverse receptor behaviors that enable highly sensitive and context-dependent hormone action. For example, in response to hormone or growth factor membrane-initiated signaling events, posttranslational modifications (PTMs) to SRs alter protein-protein interactions that govern the complex process of promoter or gene-set selection coupled to transcriptional repression or activation. Unique phosphorylation events allow SRs to associate or disassociate with specific cofactors that may include pioneer factors and other tethering partners, which specify the resulting transcriptome and ultimately change cell fate. The impact of PTMs on SR action is particularly profound in the context of breast tumorigenesis, in which frequent alterations in growth factor-initiated signaling pathways occur early and act as drivers of breast cancer progression toward endocrine resistance. In this article, with primary focus on breast cancer relevance, we review the mechanisms by which PTMs, including reversible phosphorylation events, regulate the closely related SRs, glucocorticoid receptor and progesterone receptor, allowing for precise biological responses to ever-changing hormonal stimuli.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Female
- Gene Expression
- Gene Expression Regulation, Neoplastic
- Humans
- Prognosis
- Protein Isoforms
- Protein Processing, Post-Translational
- Receptors, Estrogen/metabolism
- Receptors, Glucocorticoid/chemistry
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Receptors, Progesterone/chemistry
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Signal Transduction
- Stress, Physiological
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Katherine A Leehy
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Tarah M Regan Anderson
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Andrea R Daniel
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Carol A Lange
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| | - Julie H Ostrander
- Department of Medicine and Pharmacology University of Minnesota Twin Cities MinneapolisMinnesota, USA
| |
Collapse
|
126
|
|
127
|
Voutsadakis IA. Hormone Receptors in Serous Ovarian Carcinoma: Prognosis, Pathogenesis, and Treatment Considerations. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2016; 10:17-25. [PMID: 27053923 PMCID: PMC4814131 DOI: 10.4137/cmo.s32813] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 12/15/2022]
Abstract
A few breakthroughs have been accomplished for the treatment of ovarian cancer, the most deadly gynecologic carcinoma, in the current era of targeted oncologic treatment. The estrogen receptor was the first target of such treatments with the introduction of tamoxifen four decades ago in breast cancer therapeutics. Attempts to duplicate the success of hormonal therapies in ovarian cancer met with mixed results, which may be due to an inferior degree of hormone dependency in this cancer. Alternatively, this may be due to the failure to clearly identify the subsets of ovarian cancer with hormone sensitivity. This article reviews the expression of hormone receptors by ovarian cancer cells, the prognostic value of these expressions, and their predictive capacity for response to hormonal agents. The possible ways ahead are briefly discussed.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON, Canada.; Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
128
|
Tamura K, Naraba H, Hara T, Nakamura K, Yoshie M, Kogo H, Tachikawa E. A positive feedback loop between progesterone and microsomal prostaglandin E synthase-1-mediated PGE2 promotes production of both in mouse granulosa cells. Prostaglandins Other Lipid Mediat 2016; 123:56-62. [DOI: 10.1016/j.prostaglandins.2016.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 04/30/2016] [Accepted: 05/04/2016] [Indexed: 12/26/2022]
|
129
|
Kavlashvili T, Jia Y, Dai D, Meng X, Thiel KW, Leslie KK, Yang S. Inverse Relationship between Progesterone Receptor and Myc in Endometrial Cancer. PLoS One 2016; 11:e0148912. [PMID: 26859414 PMCID: PMC4747472 DOI: 10.1371/journal.pone.0148912] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/24/2016] [Indexed: 12/15/2022] Open
Abstract
Endometrial cancer, the most common gynecologic malignancy, is a hormonally-regulated disease. Response to progestin therapy positively correlates with hormone receptor expression, in particular progesterone receptor (PR). However, many advanced tumors lose PR expression. We recently reported that the efficacy of progestin therapy can be significantly enhanced by combining progestin with epigenetic modulators, which we term “molecularly enhanced progestin therapy.” What remained unclear was the mechanism of action and if estrogen receptor α (ERα), the principle inducer of PR, is necessary to restore functional expression of PR via molecularly enhanced progestin therapy. Therefore, we modeled advanced endometrial tumors that have lost both ERα and PR expression by generating ERα-null endometrial cancer cell lines. CRISPR-Cas9 technology was used to delete ERα at the genomic level. Our data demonstrate that treatment with a histone deacetylase inhibitor (HDACi) was sufficient to restore functional PR expression, even in cells devoid of ERα. Our studies also revealed that HDACi treatment results in marked downregulation of the oncogene Myc. We established that PR is a negative transcriptional regulator of Myc in endometrial cancer in the presence or absence of ERα, which is in contrast to studies in breast cancer cells. First, estrogen stimulation augmented PR expression and decreased Myc in endometrial cancer cell lines. Second, progesterone increased PR activity yet blunted Myc mRNA and protein expression. Finally, overexpression of PR by adenoviral transduction in ERα-null endometrial cancer cells significantly decreased expression of Myc and Myc-regulated genes. Analysis of the Cancer Genome Atlas (TCGA) database of endometrial tumors identified an inverse correlation between PR and Myc mRNA levels, with a corresponding inverse correlation between PR and Myc downstream transcriptional targets SRD5A1, CDK2 and CCNB1. Together, these data reveal a previously unanticipated inverse relationship between the tumor suppressor PR and the oncogene Myc in endometrial cancer.
Collapse
Affiliation(s)
- Tamar Kavlashvili
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, United States of America
| | - Yichen Jia
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, United States of America
| | - Donghai Dai
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, United States of America
| | - Xiangbing Meng
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, United States of America
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States of America
| | - Kristina W. Thiel
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, United States of America
| | - Kimberly K. Leslie
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, United States of America
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States of America
| | - Shujie Yang
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, United States of America
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
130
|
Voutsadakis IA. Epithelial-Mesenchymal Transition (EMT) and Regulation of EMT Factors by Steroid Nuclear Receptors in Breast Cancer: A Review and in Silico Investigation. J Clin Med 2016; 5:E11. [PMID: 26797644 PMCID: PMC4730136 DOI: 10.3390/jcm5010011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 12/23/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022] Open
Abstract
Steroid Nuclear Receptors (SNRs) are transcription factors of the nuclear receptor super-family. Estrogen Receptor (ERα) is the best-studied and has a seminal role in the clinic both as a prognostic marker but also as a predictor of response to anti-estrogenic therapies. Progesterone Receptor (PR) is also used in the clinic but with a more debatable prognostic role and the role of the four other SNRs, ERβ, Androgen Receptor (AR), Glucocorticoid Receptor (GR) and Mineralocorticoid Receptor (MR), is starting only to be appreciated. ERα, but also to a certain degree the other SNRs, have been reported to be involved in virtually every cancer-enabling process, both promoting and impeding carcinogenesis. Epithelial-Mesenchymal Transition (EMT) and the reverse Mesenchymal Epithelial Transition (MET) are such carcinogenesis-enabling processes with important roles in invasion and metastasis initiation but also establishment of tumor in the metastatic site. EMT is governed by several signal transduction pathways culminating in core transcription factors of the process, such as Snail, Slug, ZEB1 and ZEB2, and Twist, among others. This paper will discuss direct regulation of these core transcription factors by SNRs in breast cancer. Interrogation of publicly available databases for binding sites of SNRs on promoters of core EMT factors will also be included in an attempt to fill gaps where other experimental data are not available.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON P6B 0A8, Canada.
- Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, QC P3E 2C6, Canada.
| |
Collapse
|
131
|
Clark NC, Friel AM, Pru CA, Zhang L, Shioda T, Rueda BR, Peluso JJ, Pru JK. Progesterone receptor membrane component 1 promotes survival of human breast cancer cells and the growth of xenograft tumors. Cancer Biol Ther 2016; 17:262-71. [PMID: 26785864 DOI: 10.1080/15384047.2016.1139240] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Triple negative breast cancers (TNBCs) are highly aggressive and grow in response to sex steroid hormones despite lacking expression of the classical estrogen (E2) and progesterone (P4) receptors. Since P4 receptor membrane component 1 (PGRMC1) is expressed in breast cancer tumors and is known to mediate P4-induced cell survival, this study was designed to determine the expression of PGRMC1 in TNBC tumors and the involvement of PGRMC1 in regulating proliferation and survival of TNBC cells in vitro and the growth of TNBC tumors in vivo. For the latter studies, the MDA-MB-231 (MDA) cell line derived from TNBC was used. These cells express PGRMC1 but lack expression of the classical P4 receptor. A lentiviral-based shRNA approach was used to generate a stably transfected PGRMC1-deplete MDA line for comparison to the PGRMC1-intact MDA line. The present studies demonstrate that PGRMC1: 1) is expressed in TNBC cells; 2) mediates the ability of P4 to suppress TNBC cell mitosis in vitro; 3) is required for P4 to reduce the apoptotic effects of doxorubicin in vitro; and 4) facilitates TNBC tumor formation and growth in vivo. Taken together, these findings indicate that PGRMC1 plays an important role in regulating the growth and survival of TNBC cells in vitro and ultimately in the formation and development of these tumors in vivo. Thus, PGRMC1 may be a therapeutic target for TNBCs.
Collapse
Affiliation(s)
- Nicole C Clark
- a Department of Animal Sciences , School of Molecular Biosciences, Center for Reproductive Biology, Washington State University , Pullman , WA , USA
| | - Anne M Friel
- b Vincent Center for Reproductive Biology and Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School , Boston , MA , USA
| | - Cindy A Pru
- a Department of Animal Sciences , School of Molecular Biosciences, Center for Reproductive Biology, Washington State University , Pullman , WA , USA
| | - Ling Zhang
- b Vincent Center for Reproductive Biology and Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School , Boston , MA , USA
| | - Toshi Shioda
- c Massachusetts General Hospital Cancer Center and Harvard Medical School , Charlestown , MA , USA
| | - Bo R Rueda
- b Vincent Center for Reproductive Biology and Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School , Boston , MA , USA
| | - John J Peluso
- d Departments of Obstetrics and Gynecology and Cell Biology , University of Connecticut Health Center , Farmington , CT , USA
| | - James K Pru
- a Department of Animal Sciences , School of Molecular Biosciences, Center for Reproductive Biology, Washington State University , Pullman , WA , USA
| |
Collapse
|
132
|
González Ricarte M, de Castro Pérez A, Tarín JJ, Cano A. Progestogens and risk of breast cancer: a link between bone and breast? Gynecol Endocrinol 2016; 32:6-8. [PMID: 26299383 DOI: 10.3109/09513590.2015.1078304] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This article reviews the data supporting the role of receptor activator of the nuclear factor kappa (RANK) and its ligand, RANKL, in progestogen-induced breast cancer. Both experimental and clinical studies have been included. The expression of both RANK and RANKL has been described in epithelial cells of both mice and humans. Experiments of gain and loss of function in mice have shown that RANK/RANKL mediate alveologenesis during pregnancy or the estrous cycle. Moreover, the participation of the RANK/RANKL has been detected in models of breast carcinogenesis associated with progestogens-like medroxyprogesterone acetate. Recent clinical studies have found that the expression of RANK is associated with parameters of aggressiveness of the tumor.
Collapse
Affiliation(s)
- María González Ricarte
- a Service of Obstetrics and Gynecology, Hospital Clínico Universitario , Valencia , Spain
| | - Ana de Castro Pérez
- a Service of Obstetrics and Gynecology, Hospital Clínico Universitario , Valencia , Spain
| | - Juan J Tarín
- b Department of Functional Biology and Physical Anthropology , School of Biological Sciences, Burjasot Campus, University of Valencia , Valencia , Spain , and
| | - Antonio Cano
- a Service of Obstetrics and Gynecology, Hospital Clínico Universitario , Valencia , Spain
- c Department of Pediatrics , Obstetrics and Gynecology, University of Valencia , Valencia , Spain
| |
Collapse
|
133
|
Diep CH, Knutson TP, Lange CA. Active FOXO1 Is a Key Determinant of Isoform-Specific Progesterone Receptor Transactivation and Senescence Programming. Mol Cancer Res 2015; 14:141-62. [PMID: 26577046 DOI: 10.1158/1541-7786.mcr-15-0431] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/04/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED Progesterone promotes differentiation coupled to proliferation and prosurvival in the breast, but inhibits estrogen-driven growth in the reproductive tract and ovaries. Herein, it is demonstrated, using progesterone receptor (PR) isoform-specific ovarian cancer model systems, that PR-A and PR-B promote distinct gene expression profiles that differ from PR-driven genes in breast cancer cells. In ovarian cancer models, PR-A primarily regulates genes independently of progestin, while PR-B is the dominant ligand-dependent isoform. Notably, FOXO1 and the PR/FOXO1 target gene p21 (CDKN1A) are repressed by PR-A, but induced by PR-B. In the presence of progestin, PR-B, but not PR-A, robustly induced cellular senescence via FOXO1-dependent induction of p21 and p15 (CDKN2B). Chromatin immunoprecipitation (ChIP) assays performed on PR isoform-specific cells demonstrated that while each isoform is recruited to the same PRE-containing region of the p21 promoter in response to progestin, only PR-B elicits active chromatin marks. Overexpression of constitutively active FOXO1 in PR-A-expressing cells conferred robust ligand-dependent upregulation of the PR-B target genes GZMA, IGFBP1, and p21, and induced cellular senescence. In the presence of endogenous active FOXO1, PR-A was phosphorylated on Ser294 and transactivated PR-B at PR-B target genes; these events were blocked by the FOXO1 inhibitor (AS1842856). PR isoform-specific regulation of the FOXO1/p21 axis recapitulated in human primary ovarian tumor explants treated with progestin; loss of progestin sensitivity correlated with high AKT activity. IMPLICATIONS This study indicates FOXO1 as a critical component for progesterone signaling to promote cellular senescence and reveals a novel mechanism for transcription factor control of hormone sensitivity.
Collapse
Affiliation(s)
- Caroline H Diep
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Todd P Knutson
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Carol A Lange
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota. Department of Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
134
|
Esber N, Le Billan F, Resche-Rigon M, Loosfelt H, Lombès M, Chabbert-Buffet N. Ulipristal Acetate Inhibits Progesterone Receptor Isoform A-Mediated Human Breast Cancer Proliferation and BCl2-L1 Expression. PLoS One 2015; 10:e0140795. [PMID: 26474308 PMCID: PMC4608808 DOI: 10.1371/journal.pone.0140795] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022] Open
Abstract
The progesterone receptor (PR) with its isoforms and ligands are involved in breast tumorigenesis and prognosis. We aimed at analyzing the respective contribution of PR isoforms, PRA and PRB, in breast cancer cell proliferation in a new estrogen-independent cell based-model, allowing independent PR isoforms analysis. We used the bi-inducible human breast cancer cell system MDA-iPRAB. We studied the effects and molecular mechanisms of action of progesterone (P4) and ulipristal acetate (UPA), a new selective progesterone receptor modulator, alone or in combination. P4 significantly stimulated MDA-iPRA expressing cells proliferation. This was associated with P4-stimulated expression of the anti-apoptotic factor BCL2-L1 and enhanced recruitment of PRA, SRC-1 and RNA Pol II onto the +58 kb PR binding motif of the BCL2-L1 gene. UPA decreased cell proliferation and repressed BCL2-L1 expression in the presence of PRA, correlating with PRA and SRC1 but not RNA Pol II recruitment. These results bring new information on the mechanism of action of PR ligands in controlling breast cancer cell proliferation through PRA in an estrogen independent model. Evaluation of PR isoforms ratio, as well as molecular signature studies based on PRA target genes could be proposed to facilitate personalized breast cancer therapy. In this context, UPA could be of interest in endocrine therapy. Further confirmation in the clinical setting is required.
Collapse
Affiliation(s)
- Nathalie Esber
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-Scientifique 1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Université Paris-Sud, Faculté de Médecine Paris Sud, Unité Mixte de Recherche-Scientifique 1185, Le Kremlin-Bicêtre, France
- HRA-Pharma, Paris, France
| | - Florian Le Billan
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-Scientifique 1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Université Paris-Sud, Faculté de Médecine Paris Sud, Unité Mixte de Recherche-Scientifique 1185, Le Kremlin-Bicêtre, France
| | | | - Hugues Loosfelt
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-Scientifique 1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Université Paris-Sud, Faculté de Médecine Paris Sud, Unité Mixte de Recherche-Scientifique 1185, Le Kremlin-Bicêtre, France
| | - Marc Lombès
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-Scientifique 1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Université Paris-Sud, Faculté de Médecine Paris Sud, Unité Mixte de Recherche-Scientifique 1185, Le Kremlin-Bicêtre, France
- Service d’Endocrinologie et des Maladies de la Reproduction, assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Nathalie Chabbert-Buffet
- Service de Gynécologie Obstétrique Médecine de la Reproduction, Hôpitaux Universitaires Est Parisien site Tenon, AP-HP, Paris, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-Scientifique 938, Centre de Recherche Saint Antoine, Université Pierre et Marie Curie, Paris, France
- * E-mail:
| |
Collapse
|
135
|
Gao K, Jin X, Tang Y, Ma J, Peng J, Yu L, Zhang P, Wang C. Tumor suppressor SPOP mediates the proteasomal degradation of progesterone receptors (PRs) in breast cancer cells. Am J Cancer Res 2015; 5:3210-3220. [PMID: 26693071 PMCID: PMC4656742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 09/03/2015] [Indexed: 06/05/2023] Open
Abstract
Progesterone induces proliferation of breast cancer cells and contributes to the development of breast cancer. The effects of progesterone are mediated by progesterone receptors (PRs). However, it is still not fully understood how the proliferative effects of PR is regulated in vivo. Increasing amount of evidence strongly suggests that dysregulation of ubiquitin-proteasome system is closely associated with cancer pathogenesis. Speckle-type POZ protein (SPOP) is an adaptor protein of the CUL3-based E3 ubiquitin ligase complexes. SPOP represents one of the highest loci for loss of heterozygosity (LOH) in breast cancer. SPOP downregulation contributes to breast cancer cell growth and invasion. In this study, we revealed PR as a bona fide substrate for SPOP. SPOP interacts with PR in vivo and targets PR for ubiquitin-dependent proteasomal degradation. Moreover, SPOP suppresses progesteroneinduced PR transactivation, S phase entry, and Erk1/2 activation. Our study revealed novel molecular mechanisms underlying the regulation of PR protein homeostasis in breast cancer cells, and provided insights in understanding the relationship between SPOP inactivation and the development of breast cancer.
Collapse
Affiliation(s)
- Kun Gao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan UniversityShanghai, P. R. China
| | - Xiaofeng Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan UniversityShanghai, P. R. China
| | - Yan Tang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan UniversityShanghai, P. R. China
| | - Jian Ma
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong UniversityShanghai, P. R. China
| | - Jingtiao Peng
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong UniversityShanghai, P. R. China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan UniversityShanghai, P. R. China
| | - Pingzhao Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan UniversityShanghai, P. R. China
| | - Chenji Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan UniversityShanghai, P. R. China
| |
Collapse
|
136
|
Zhang P, Wang C, Cheng L, Zhang P, Guo L, Liu W, Zhang Z, Huang Y, Ou Q, Wen X, Tian Y. Development of a multi-marker model combining HE4, CA125, progesterone, and estradiol for distinguishing benign from malignant pelvic masses in postmenopausal women. Tumour Biol 2015; 37:2183-91. [DOI: 10.1007/s13277-015-4037-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/02/2015] [Indexed: 01/04/2023] Open
|
137
|
Koizume S, Miyagi Y. Tissue Factor-Factor VII Complex As a Key Regulator of Ovarian Cancer Phenotypes. BIOMARKERS IN CANCER 2015; 7:1-13. [PMID: 26396550 PMCID: PMC4562604 DOI: 10.4137/bic.s29318] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/05/2015] [Accepted: 08/07/2015] [Indexed: 02/07/2023]
Abstract
Tissue factor (TF) is an integral membrane protein widely expressed in normal human cells. Blood coagulation factor VII (fVII) is a key enzyme in the extrinsic coagulation cascade that is predominantly secreted by hepatocytes and released into the bloodstream. The TF–fVII complex is aberrantly expressed on the surface of cancer cells, including ovarian cancer cells. This procoagulant complex can initiate intracellular signaling mechanisms, resulting in malignant phenotypes. Cancer tissues are chronically exposed to hypoxia. TF and fVII can be induced in response to hypoxia in ovarian cancer cells at the gene expression level, leading to the autonomous production of the TF–fVII complex. Here, we discuss the roles of the TF–fVII complex in the induction of malignant phenotypes in ovarian cancer cells. The hypoxic nature of ovarian cancer tissues and the roles of TF expression in endometriosis are discussed. Arguments will be extended to potential strategies to treat ovarian cancers based on our current knowledge of TF–fVII function.
Collapse
Affiliation(s)
- Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|